Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Minhong Yan is active.

Publication


Featured researches published by Minhong Yan.


Journal of Clinical Investigation | 2013

Blockade of individual Notch ligands and receptors controls graft-versus-host disease

Ivy T. Tran; Ashley R. Sandy; Alexis J. Carulli; Christen L. Ebens; Jooho Chung; Gloria T. Shan; Vedran Radojcic; Ann Friedman; Thomas Gridley; Amy Shelton; Pavan Reddy; Linda C. Samuelson; Minhong Yan; Christian W. Siebel; Ivan Maillard

Graft-versus-host disease (GVHD) is the main complication of allogeneic bone marrow transplantation. Current strategies to control GVHD rely on global immunosuppression. These strategies are incompletely effective and decrease the anticancer activity of the allogeneic graft. We previously identified Notch signaling in T cells as a new therapeutic target for preventing GVHD. Notch-deprived T cells showed markedly decreased production of inflammatory cytokines, but normal in vivo proliferation, increased accumulation of regulatory T cells, and preserved anticancer effects. Here, we report that γ-secretase inhibitors can block all Notch signals in alloreactive T cells, but lead to severe on-target intestinal toxicity. Using newly developed humanized antibodies and conditional genetic models, we demonstrate that Notch1/Notch2 receptors and the Notch ligands Delta-like1/4 mediate all the effects of Notch signaling in T cells during GVHD, with dominant roles for Notch1 and Delta-like4. Notch1 inhibition controlled GVHD, but led to treatment-limiting toxicity. In contrast, Delta-like1/4 inhibition blocked GVHD without limiting adverse effects while preserving substantial anticancer activity. Transient blockade in the peritransplant period provided durable protection. These findings open new perspectives for selective and safe targeting of individual Notch pathway components in GVHD and other T cell-mediated human disorders.


Blood | 2010

ALK1 signaling regulates early postnatal lymphatic vessel development

Kyle Niessen; Gu Zhang; John Ridgway; Hao Chen; Minhong Yan

In vertebrates, endothelial cells form 2 hierarchical tubular networks, the blood vessels and the lymphatic vessels. Despite the difference in their structure and function and genetic programs that dictate their morphogenesis, common signaling pathways have been recognized that regulate both vascular systems. ALK1 is a member of the transforming growth factor-beta type I family of receptors, and compelling genetic evidence suggests its essential role in regulating blood vascular development. Here we report that ALK1 signaling is intimately involved in lymphatic development. Lymphatic endothelial cells express key components of the ALK1 pathway and respond robustly to ALK1 ligand stimulation in vitro. Blockade of ALK1 signaling results in defective lymphatic development in multiple organs of neonatal mice. We find that ALK1 signaling regulates the differentiation of lymphatic endothelial cells to influence the lymphatic vascular development and remodeling. Furthermore, simultaneous inhibition of ALK1 pathway increases apoptosis in lymphatic vessels caused by blockade of VEGFR3 signaling. Thus, our study reveals a novel aspect of ALK1 signaling in regulating lymphatic development and suggests that targeting ALK1 pathway might provide additional control of lymphangiogenesis in human diseases.


Nature Communications | 2015

Notch signal strength controls cell fate in the haemogenic endothelium.

Leonor Gama-Norton; Eva Ferrando; Cristina Ruiz-Herguido; Zhenyi Liu; Jordi Guiu; Abul B.M.M.K. Islam; Sung-Uk Lee; Minhong Yan; Cynthia J. Guidos; Nuria Lopez-Bigas; Takahiro Maeda; Lluis Espinosa; Raphael Kopan; Anna Bigas

Acquisition of the arterial and haemogenic endothelium fates concurrently occur in the aorta–gonad–mesonephros (AGM) region prior to haematopoietic stem cell (HSC) generation. The arterial programme depends on Dll4 and the haemogenic endothelium/HSC on Jag1-mediated Notch1 signalling. How Notch1 distinguishes and executes these different programmes in response to particular ligands is poorly understood. By using two Notch1 activation trap mouse models with different sensitivity, here we show that arterial endothelial cells and HSCs originate from distinct precursors, characterized by different Notch1 signal strengths. Microarray analysis on AGM subpopulations demonstrates that the Jag1 ligand stimulates low Notch strength, inhibits the endothelial programme and is permissive for HSC specification. In the absence of Jag1, endothelial cells experience high Dll4-induced Notch activity and select the endothelial programme, thus precluding HSC formation. Interference with the Dll4 signal by ligand-specific blocking antibodies is sufficient to inhibit the endothelial programme and favour specification of the haematopoietic lineage.


Blood | 2013

LRF-mediated Dll4 repression in erythroblasts is necessary for hematopoietic stem cell maintenance

Sung Uk Lee; Manami Maeda; Yuichi Ishikawa; Sierra Min Li; Anne Wilson; Adrian M. Jubb; Nagisa Sakurai; Lihong Weng; Emma Fiorini; Freddy Radtke; Minhong Yan; H. Robson MacDonald; Ching Cheng Chen; Takahiro Maeda

Hematopoietic stem cells (HSCs) are the most primitive cells in the hematopoietic system and are under tight regulation for self-renewal and differentiation. Notch signals are essential for the emergence of definitive hematopoiesis in mouse embryos and are critical regulators of lymphoid lineage fate determination. However, it remains unclear how Notch regulates the balance between HSC self-renewal and differentiation in the adult bone marrow (BM). Here we report a novel mechanism that prevents HSCs from undergoing premature lymphoid differentiation in BM. Using a series of in vivo mouse models and functional HSC assays, we show that leukemia/lymphoma related factor (LRF) is necessary for HSC maintenance by functioning as an erythroid-specific repressor of Delta-like 4 (Dll4) expression. Lrf deletion in erythroblasts promoted up-regulation of Dll4 in erythroblasts, sensitizing HSCs to T-cell instructive signals in the BM. Our study reveals novel cross-talk between HSCs and erythroblasts, and sheds a new light on the regulatory mechanisms regulating the balance between HSC self-renewal and differentiation.


Stem Cells | 2015

Notch Receptor‐Ligand Engagement Maintains Hematopoietic Stem Cell Quiescence and Niche Retention

Weihuan Wang; Shuiliang Yu; Grant Zimmerman; Yiwei Wang; Jay Myers; Vionnie W.C. Yu; Dan Huang; Xiaoran Huang; Jeongsup Shim; Y. Huang; William W. Xin; Peter Qiao; Minhong Yan; Wei Xin; David T. Scadden; Pamela Stanley; John B. Lowe; Alex Y. Huang; Christian W. Siebel; Lan Zhou

Notch is long recognized as a signaling molecule important for stem cell self‐renewal and fate determination. Here, we reveal a novel adhesive role of Notch‐ligand engagement in hematopoietic stem and progenitor cells (HSPCs). Using mice with conditional loss of O‐fucosylglycans on Notch EGF‐like repeats important for the binding of Notch ligands, we report that HSPCs with faulty ligand binding ability display enhanced cycling accompanied by increased egress from the marrow, a phenotype mainly attributed to their reduced adhesion to Notch ligand‐expressing stromal cells and osteoblastic cells and their altered occupation in osteoblastic niches. Adhesion to Notch ligand‐bearing osteoblastic or stromal cells inhibits wild type but not O‐fucosylglycan‐deficient HSPC cycling, independent of RBP‐JK‐mediated canonical Notch signaling. Furthermore, Notch‐ligand neutralizing antibodies induce RBP‐JK‐independent HSPC egress and enhanced HSPC mobilization. We, therefore, conclude that Notch receptor–ligand engagement controls HSPC quiescence and retention in the marrow niche that is dependent on O‐fucosylglycans on Notch. Stem Cells 2015;33:2280–2293


Journal of Immunology | 2015

Transient Blockade of Delta-like Notch Ligands Prevents Allograft Rejection Mediated by Cellular and Humoral Mechanisms in a Mouse Model of Heart Transplantation

Sherri C. Wood; Jiane Feng; Jooho Chung; Vedran Radojcic; Ashley R. Sandy-Sloat; Ann Friedman; Amy Shelton; Minhong Yan; Christian W. Siebel; D. Keith Bishop; Ivan Maillard

Rejection remains a major clinical challenge limiting allograft survival after solid organ transplantation. Both cellular and humoral immunity contribute to this complication, with increased recognition of Ab-mediated damage during acute and chronic rejection. Using a mouse model of MHC-mismatched heart transplantation, we report markedly protective effects of Notch inhibition, dampening both T cell and Ab-driven rejection. T cell–specific pan-Notch blockade prolonged heart allograft survival and decreased IFN-γ and IL-4 production by alloreactive T cells, especially when combined with depletion of recipient CD8+ T cells. These effects were associated with decreased infiltration by conventional T cells and an increased proportion of regulatory T cells in the graft. Transient administration of neutralizing Abs specific for delta-like (Dll)1/4 Notch ligands in the peritransplant period led to prolonged acceptance of allogeneic hearts, with superior outcome over Notch inhibition only in T cells. Systemic Dll1/4 inhibition decreased T cell cytokines and graft infiltration, germinal center B cell and plasmablast numbers, as well as production of donor-specific alloantibodies and complement deposition in the transplanted hearts. Dll1 or Dll4 inhibition alone provided partial protection. Thus, pathogenic signals delivered by Dll1/4 Notch ligands early after transplantation promote organ rejection through several complementary mechanisms. Transient interruption of these signals represents an attractive new therapeutic strategy to enhance long-term allograft survival.


Journal of Clinical Investigation | 2017

Fibroblastic niches prime T cell alloimmunity through Delta-like Notch ligands

Jooho Chung; Christen L. Ebens; Eric Perkey; Vedran Radojcic; Ute Koch; Leonardo Scarpellino; Alexander Tong; Frederick Allen; Sherri C. Wood; Jiane Feng; Ann Friedman; David Granadier; Ivy T. Tran; Qian Chai; Lucas Onder; Minhong Yan; Pavan Reddy; Bruce R. Blazar; Alex Y. Huang; Todd V. Brennan; D. Keith Bishop; Burkhard Ludewig; Christian W. Siebel; Freddy Radtke; Sanjiv A. Luther; Ivan Maillard

Alloimmune T cell responses induce graft-versus-host disease (GVHD), a serious complication of allogeneic bone marrow transplantation (allo-BMT). Although Notch signaling mediated by Delta-like 1/4 (DLL1/4) Notch ligands has emerged as a major regulator of GVHD pathogenesis, little is known about the timing of essential Notch signals and the cellular source of Notch ligands after allo-BMT. Here, we have shown that critical DLL1/4-mediated Notch signals are delivered to donor T cells during a short 48-hour window after transplantation in a mouse allo-BMT model. Stromal, but not hematopoietic, cells were the essential source of Notch ligands during in vivo priming of alloreactive T cells. GVHD could be prevented by selective inactivation of Dll1 and Dll4 in subsets of fibroblastic stromal cells that were derived from chemokine Ccl19-expressing host cells, including fibroblastic reticular cells and follicular dendritic cells. However, neither T cell recruitment into secondary lymphoid organs nor initial T cell activation was affected by Dll1/4 loss. Thus, we have uncovered a pathogenic function for fibroblastic stromal cells in alloimmune reactivity that can be dissociated from their homeostatic functions. Our results reveal what we believe to be a previously unrecognized Notch-mediated immunopathogenic role for stromal cell niches in secondary lymphoid organs after allo-BMT and define a framework of early cellular and molecular interactions that regulate T cell alloimmunity.


Clinical Cancer Research | 2016

Balancing efficacy and safety of an anti-DLL4 antibody through pharmacokinetic modulation

Jessica Couch; Gu Zhang; Joseph Beyer; Christina L.Zuch de Zafra; Priyanka Gupta; Amrita V. Kamath; Nicholas Lewin-Koh; Jacqueline M. Tarrant; Krishna P. Allamneni; Gary Cain; Sharon Yee; Sarajane Ross; Ryan Cook; Siao Ping Tsai; Jane Ruppel; John Ridgway; Maciej Paluch; Philip E. Hass; Jayme Franklin; Minhong Yan

Purpose: Although agents targeting Delta-like ligand 4 (DLL4) have shown great promise for angiogenesis-based cancer therapy, findings in recent studies have raised serious safety concerns. To further evaluate the potential for therapeutic targeting of the DLL4 pathway, we pursued a novel strategy to reduce toxicities related to DLL4 inhibition by modulating the pharmacokinetic (PK) properties of an anti-DLL4 antibody. Experimental Design: The F(ab′)2 fragment of anti-DLL4 antibody (anti-DLL4 F(ab′)2) was generated and assessed in efficacy and toxicity studies. Results: Anti-DLL4 F(ab′)2 enables greater control over the extent and duration of DLL4 inhibition, such that intermittent dosing of anti-DLL4 F(ab′)2 can maintain significant antitumor activity while markedly mitigating known toxicities associated with continuous pathway inhibition. Conclusions: PK modulation has potentially broad implications for development of antibody-based therapeutics. Our safety studies with anti-DLL4 F(ab′)2 also provide new evidence reinforcing the notion that the DLL4 pathway is extremely sensitive to pharmacologic perturbation, further underscoring the importance of exercising caution to safely harness this potent pathway in humans. Clin Cancer Res; 22(6); 1469–79. ©2015 AACR.


Nature Communications | 2016

Corrigendum: Notch signal strength controls cell fate in the haemogenic endothelium

Leonor Gama-Norton; Eva Ferrando; Cristina Ruiz-Herguido; Zhenyi Liu; Jordi Guiu; Abul B.M.M.K. Islam; Sung-Uk Lee; Minhong Yan; Cynthia J. Guidos; Nuria Lopez-Bigas; Takahiro Maeda; Lluis Espinosa; Raphael Kopan; Anna Bigas

Nature Communications 6: Article number: 8510 10.1038/ncomms9510 (2015); Published: October142015; Updated: March042016 n nThe original version of this Article contained a typographical error in the spelling of the author Zhenyi Liu, which was incorrectly given as Zenhy Liu. This has now been corrected in both the PDF and HTML versions of the Article.


Blood | 2011

In Vivo Blockade of Individual Notch Ligands and Receptors Provides a New Targeted Therapeutic Approach In Graft-Versus-Host Disease

Ivy T. Tran; Ashley R. Sandy; Alexis J. Carulli; Gloria T. Shan; Vedran Radojcic; Ann Friedman; Amy Shelton; Linda C. Samuelson; Minhong Yan; Christian W. Siebel; Ivan Maillard

Collaboration


Dive into the Minhong Yan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sung-Uk Lee

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manami Maeda

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Freddy Radtke

École Polytechnique Fédérale de Lausanne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alex Y. Huang

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge